1
|
Su W, Liang Z, Pan D, Zhang L, Zhang Y, Yuan T, Gao X, Su H, Zhang H. Therapeutic effect of notoginseng saponins before and after fermentation on blood deficiency rats. Exp Ther Med 2024; 27:143. [PMID: 38476921 PMCID: PMC10928825 DOI: 10.3892/etm.2024.12431] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 01/12/2024] [Indexed: 03/14/2024] Open
Abstract
Notoginseng saponins (NS) are the active ingredients in Panax notoginseng (Burk.) F.H. Chen (PN). NS can be transformed depending on how the extract is processed. Fermentation has been shown to produce secondary ginsenosides with increased bioavailability. However, the therapeutic effect of fermented NS (FNS) requires further study. The present study compared the compositions and activities of FNS and NS in blood deficiency rats, which resembles the symptoms of anemia in modern medicine, induced by acetylphenylhydrazine and cyclophosphamide. A total of 32 rats were randomly divided into control, model, FNS and NS groups. A blood deficiency model was established and then treatment was orally administered for 21 days. The results of component analysis indicated that some saponins transformed during the fermentation process resulting in a decrease of notoginsenoside R1, and ginsenosides Rg1, Rb1 and Re, and an increase in ginsenosides Rd, Rh2, compound K, protopanaxadiol and protopanaxatriol. The animal results showed that both FNS and NS increased the number of white blood cells (WBCs), red blood cells, hemoglobin, platelets and reticulocytes, and the levels of granulocyte-macrophage colony-stimulating factor (GM-CSF), erythropoietin (EPO) and thrombopoietin (TPO), decreased the G0/G1 phase and increased G2/M phase, and decreased the apoptosis rate of bone marrow (BM) cells, which suggested a contribution to the recovery of hematopoietic function of the BM cells. FNS and NS increased the protein expression levels of the cytokines IL-4, IL-10, IL-12, IL-13, TGF-β, IL-6, IFN-γ and TNF-α, and the mRNA expression levels of transcription factors GATA binding protein 3 and T-box expressed in T cell (T-bet). FNS and NS treatment also increased the number of CD4+ T cells, and decreased the enlargement of the rat spleen and thymus atrophy, which indicated a protective effect on the organs of the immune system. The results of the present study demonstrated that compared with NS, FNS showed an improved ability to increase the levels of WBCs, lymphocytes, GM-CSF, EPO, TPO, aspartate aminotransferase, IL-10, IL-12, IL-13 and TNF-α, and the mRNA expression levels of T-bet, and decrease alanine aminotransferase levels. The differences seen for FNS treatment could arise from their improved bioavailability compared with NS, due to the larger proportion of hydrophobic ginsenosides produced during fermentation.
Collapse
Affiliation(s)
- Wenjie Su
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin 130021, P.R. China
| | - Zuguo Liang
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Daian Pan
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin 130021, P.R. China
| | - Lancao Zhang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Yuyao Zhang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Tongyi Yuan
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Xiang Gao
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Hang Su
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - He Zhang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
2
|
Role of myeloid-derived suppressor cells in tumor recurrence. Cancer Metastasis Rev 2023; 42:113-142. [PMID: 36640224 PMCID: PMC9840433 DOI: 10.1007/s10555-023-10079-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
The establishment of primary tumor cells in distant organs, termed metastasis, is the principal cause of cancer mortality and is a crucial therapeutic target in oncology. Thus, it is critical to establish a better understanding of metastatic progression for the future development of improved therapeutic approaches. Indeed, such development requires insight into the timing of tumor cell dissemination and seeding of distant organs resulting in occult lesions. Following dissemination of tumor cells from the primary tumor, they can reside in niches in distant organs for years or decades, following which they can emerge as an overt metastasis. This timeline of metastatic dormancy is regulated by interactions between the tumor, its microenvironment, angiogenesis, and tumor antigen-specific T-cell responses. An improved understanding of the mechanisms and interactions responsible for immune evasion and tumor cell release from dormancy would help identify and aid in the development of novel targeted therapeutics. One such mediator of dormancy is myeloid derived suppressor cells (MDSC), whose number in the peripheral blood (PB) or infiltrating tumors has been associated with cancer stage, grade, patient survival, and metastasis in a broad range of tumor pathologies. Thus, extensive studies have revealed a role for MDSCs in tumor escape from adoptive and innate immune responses, facilitating tumor progression and metastasis; however, few studies have considered their role in dormancy. We have posited that MDSCs may regulate disseminated tumor cells resulting in resurgence of senescent tumor cells. In this review, we discuss clinical studies that address mechanisms of tumor recurrence including from dormancy, the role of MDSCs in their escape from dormancy during recurrence, the development of occult metastases, and the potential for MDSC inhibition as an approach to prolong the survival of patients with advanced malignancies. We stress that assessing the impact of therapies on MDSCs versus other cellular targets is challenging within the multimodality interventions required clinically.
Collapse
|
3
|
Wang J, Wang F, Yuan L, Ruan H, Zhu Z, Fan X, Zhu L, Peng X. Blood-Enriching Effects and Immune-Regulation Mechanism of Steam-Processed Polygonatum Sibiricum Polysaccharide in Blood Deficiency Syndrome Mice. Front Immunol 2022; 13:813676. [PMID: 35250989 PMCID: PMC8892585 DOI: 10.3389/fimmu.2022.813676] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/31/2022] [Indexed: 12/13/2022] Open
Abstract
Polygonatum sibiricum Red. has been used as a medicinal herb and nutritional food in traditional Chinese medicine for a long time. It must be processed prior to clinical use for safe and effective applications. However, the present studies mainly focused on crude Polygonatum sibiricum (PS). This study aimed to investigate the chemical properties, blood-enriching effects and mechanism of polysaccharide from the steam-processed Polygonatum sibiricum (SPS), which is a common form of PS in clinical applications. Instrumentation analyses and chemistry analyses revealed the structure of SPS polysaccharide (SPSP). A mice model of blood deficiency syndrome (BDS) was induced by acetylphenylhydrazine (APH) and cyclophosphamide (CTX). Blood routine test, spleen histopathological changes, serum cytokines, etc. were measured. The spleen transcriptome changes of BDS mice were detected by RNA sequencing (RNA-seq). The results showed that SPSP consists predominantly of Gal and GalA together with fewer amounts of Man, Glc, Ara, Rha and GlcN. It could significantly increase peripheral blood cells, restore the splenic trabecular structure, and reverse hematopoietic cytokines to normal levels. RNA-seq analysis showed that 122 differentially expressed genes (DEGs) were obtained after SPSP treatment. GO and KEGG analysis revealed that SPSP-regulated DEGs were mainly involved in hematopoiesis, immune regulation signaling pathways. The reliability of transcriptome profiling was validated by quantitative real-time PCR and Western blot, and the results indicated that the potential molecular mechanisms of the blood-enriching effects of SPSP might be associated with the regulating of JAK1-STAT1 pathway, and elevated the hematopoietic cytokines (EPO, G-CSF, TNF-α and IL-6). This work provides important information on the potential mechanisms of SPSP against BDS.
Collapse
Affiliation(s)
- Juan Wang
- Department of Traditional Chinese Medicine, Zhejiang Pharmaceutical College, Ningbo, China
| | - Furong Wang
- Department of Traditional Chinese Medicine, Zhejiang Pharmaceutical College, Ningbo, China
| | - Lixia Yuan
- Department of Traditional Chinese Medicine, Zhejiang Pharmaceutical College, Ningbo, China
| | - Hongsheng Ruan
- Department of Traditional Chinese Medicine, Zhejiang Pharmaceutical College, Ningbo, China
| | - Zhibiao Zhu
- Department of Quality Control, Zhejiang Sanxitang Chinese Medicine Co., LTD, Yiwu, China
| | - Xiaoling Fan
- Department of Quality Control, Zhejiang Sanxitang Chinese Medicine Co., LTD, Yiwu, China
| | - Lingyan Zhu
- Department of Quality Control, Zhejiang Sanxitang Chinese Medicine Co., LTD, Yiwu, China
| | - Xin Peng
- Ningbo Research Institute of Zhejiang University, Ningbo, China
| |
Collapse
|
4
|
Sultan S. Aberrant expression of proatherogenic cytokines and growth factors in human umbilical vein endothelial cells from newborns of type 2 diabetic women. SAGE Open Med 2021; 9:20503121211026832. [PMID: 34211712 PMCID: PMC8216400 DOI: 10.1177/20503121211026832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/02/2021] [Indexed: 11/17/2022] Open
Abstract
Objectives: This study reports the levels of cytokines, chemokines, and growth factors previously identified as taking part in the pathology of atherosclerosis in human umbilical vein endothelial cells derived from mothers with type 2 diabetes and compares them with those in human umbilical vein endothelial cells derived from healthy mothers under normal glucose conditions. Methods: Cytokine analysis measures of human umbilical vein endothelial cell lysates were obtained using a multiple analyte profiling (xMAP) assay based on magnetic bead-based technology, using the MAGPIX instrument. The correlation between cytokines, chemokines, and growth factors was examined statistically in human umbilical vein endothelial cells derived from mothers with type 2 diabetes. Results: This study showed that the expression of proinflammatory cytokine interleukin-1 alpha was significantly greater in human umbilical vein endothelial cells derived from mothers with type 2 diabetes than those derived from healthy mothers. The protein level of granulocyte colony-stimulating factor was higher in human umbilical vein endothelial cells derived from mothers with type 2 diabetes than those derived from healthy mothers. A significant positive correlation was demonstrated between the protein expression of granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor in human umbilical vein endothelial cells derived from mothers with type 2 diabetes. Conclusion: Diabetes evokes a persistent inflammatory phenotype in human umbilical vein endothelial cells, as indicated by the enhanced production of cytokines and growth factors under normal glucose conditions.
Collapse
Affiliation(s)
- Samar Sultan
- Medical Laboratory Technology Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
5
|
Khadge S, Cole K, Talmadge JE. Myeloid derived suppressor cells and the release of micro-metastases from dormancy. Clin Exp Metastasis 2021; 38:279-293. [PMID: 34014424 DOI: 10.1007/s10585-021-10098-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/22/2021] [Indexed: 12/11/2022]
Abstract
Metastasis is the primary cause of cancer mortality and an improved understanding of its pathology is critical to the development of novel therapeutic approaches. Mechanism-based therapeutic strategies require insight into the timing of tumor cell dissemination, seeding of distant organs, formation of occult lesions and critically, their release from dormancy. Due to imaging limitations, primary tumors can only be detected when they reach a relatively large size (e.g. > 1 cm3), which, based on our understanding of tumor evolution, occurs approximately 10 years and about 30 doubling times following tumor initiation. Genomic profiling of paired primary tumors and metastases has suggested that tumor seeding at secondary sites occurs early during tumor progression and frequently, years prior to clinical diagnosis. Following seeding, tumor cells may enter into and remain in a dormant state, and if they survive and are released from dormancy, they can proliferate into an overt lesion. The timeline of tumor initiation and metastatic dormancy is regulated by tumor interactions with its microenvironment, angiogenesis, and tumor-specific cytotoxic T-lymphocyte (CTL) responses. Therefore, a better understanding of the cellular interactions responsible for immune evasion and/or tumor cell release from dormancy would facilitate the development of therapeutics targeted against this critical part of tumor progression. The immunosuppressive mechanisms mediated by myeloid-derived suppressor cells (MDSCs) contribute to tumor progression and, we posit, promote tumor cell escape from CTL-associated dormancy. Thus, while clinical and translational research has demonstrated a role for MDSCs in facilitating tumor progression and metastasis through tumor escape from adoptive and innate immune responses (T-, natural killer and B-cell responses), few studies have considered the role of MDSCs in tumor release from dormancy. In this review, we discuss MDSC expansion, driven by tumor burden associated growth factor secretion and their role in tumor cell escape from dormancy, resulting in manifest metastases. Thus, the therapeutic strategies to inhibit MDSC expansion and function may provide an approach to delay metastatic relapse and prolong the survival of patients with advanced malignancies.
Collapse
Affiliation(s)
- Saraswoti Khadge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kathryn Cole
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - James E Talmadge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198-5950, USA. .,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-6495, USA.
| |
Collapse
|
6
|
Role of myeloid-derived suppressor cells in metastasis. Cancer Metastasis Rev 2021; 40:391-411. [PMID: 33411082 DOI: 10.1007/s10555-020-09947-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 12/02/2020] [Indexed: 02/06/2023]
Abstract
The spread of primary tumor cells to distant organs, termed metastasis, is the principal cause of cancer mortality and is a critical therapeutic target in oncology. Thus, a better understanding of metastatic progression is critical for improved therapeutic approaches requiring insight into the timing of tumor cell dissemination and seeding of distant organs, which can lead to the formation of occult lesions. However, due to limitations in imaging techniques, primary tumors can only be detected when they reach a relatively large size (e.g., > 1 cm3), which, based on our understanding of tumor evolution, is 10 to 20 years (30 doubling times) following tumor initiation. Recent insights into the timing of metastasis are based on the genomic profiling of paired primary tumors and metastases, suggesting that tumor cell seeding of secondary sites occurs early during tumor progression and years prior to diagnosis. Following seeding, tumor cells may remain in a dormant state as single cells or micrometastases before emerging as overt lesions. This timeline and the role of metastatic dormancy are regulated by interactions between the tumor, its microenvironment, and tumor-specific T cell responses. An improved understanding of the mechanisms and interactions responsible for immune evasion and tumor cell release from dormancy would support the development of novel targeted therapeutics. We posit herein that the immunosuppressive mechanisms mediated by myeloid-derived suppressor cells (MDSCs) are a major contributor to tumor progression, and that these mechanisms promote tumor cell escape from dormancy. Thus, while extensive studies have demonstrated a role for MDSCs in the escape from adoptive and innate immune responses (T-, natural killer (NK)-, and B cell responses), facilitating tumor progression and metastasis, few studies have considered their role in dormancy. In this review, we discuss the role of MDSC expansion, driven by tumor burden, and its role in escape from dormancy, resulting in occult metastases, and the potential for MDSC inhibition as an approach to prolong the survival of patients with advanced malignancies.
Collapse
|
7
|
Lewis S, Holmes C. Host Defense Mechanisms in the Peritoneal Cavity of Continuous Ambulatory Peritoneal Dialysis Patients: First of Two Parts. Perit Dial Int 2020. [DOI: 10.1177/089686089101100105] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
This article provides a review of studies on peritoneal white blood cells (WBC) in CAPD patients. To some extent these studies support the concept that the peritoneal cavity of these patients contains adequate-functioning WBC that can provide effective antimicrobial defenses when they are studied in dialysate-free media. Commercially available dialysis solutions significantly impair WBC function. In some patients with high incidences of peritonitis, there appears to be reduced bactericidal capacity of their peritoneal macrophages. CAPD seems to contribute to a state of both macrophage and lymphocyte activation in the peritoneal cavity. The clinical consequences of this chronic activation are not known.
Collapse
Affiliation(s)
- Sharon Lewis
- Department of Pathology, BRF #323, University of New Mexico, School of Medicine, Albuquerque, NM
| | | |
Collapse
|
8
|
Zhang Z, Zhang Y, Gao M, Cui X, Yang Y, van Duijn B, Wang M, Hu Y, Wang C, Xiong Y. Steamed Panax notoginseng Attenuates Anemia in Mice With Blood Deficiency Syndrome via Regulating Hematopoietic Factors and JAK-STAT Pathway. Front Pharmacol 2020; 10:1578. [PMID: 32038252 PMCID: PMC6985777 DOI: 10.3389/fphar.2019.01578] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 12/05/2019] [Indexed: 12/13/2022] Open
Abstract
Panax notoginseng (Burk.) F. H. Chen is a medicinal herb used to treat blood disorders since ancient times, of which the steamed form exhibits the anti-anemia effect and acts with a “blood-tonifying” function according to traditional use. The present study aimed to investigate the anti-anemia effect and underlying mechanism of steamed P. notoginseng (SPN) on mice with blood deficiency syndrome induced by chemotherapy. Blood deficiency syndrome was induced in mice by cyclophosphamide and acetylphenylhydrazine. A number of peripheral blood cells and organs (liver, kidney, and spleen) coefficients were measured. The mRNA expression of hematopoietic function-related cytokines in the bone marrow of mice was detected by RT-qPCR. The janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathway was screened based on our previous analysis by network pharmacology. The expression of related proteins and cell cycle factors predicted in the pathway was determined by Western blot and RT-qPCR. SPN could significantly increase the numbers of peripheral blood cells and reverse the enlargement of spleen in a dose-dependent manner. The quantities of related hematopoietic factors in bone marrow were also increased significantly after SPN administration. SPN was involved in the cell cycle reaction and activation of immune cells through the JAK-STAT pathway, which could promote the hematopoiesis.
Collapse
Affiliation(s)
- Zejun Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Yiming Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Min Gao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Xiuming Cui
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Yang Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Bert van Duijn
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands.,Fytagoras BV, Leiden, Netherlands
| | - Mei Wang
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands.,LU-European Center for Chinese Medicine, Leiden University, Leiden, Netherlands.,SUBioMedicine BV, Leiden, Netherlands
| | - Yupiao Hu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Chengxiao Wang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Yin Xiong
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China.,Institute of Biology Leiden, Leiden University, Leiden, Netherlands.,Fytagoras BV, Leiden, Netherlands.,LU-European Center for Chinese Medicine, Leiden University, Leiden, Netherlands
| |
Collapse
|
9
|
Hasselmann J, Blurton-Jones M. Human iPSC-derived microglia: A growing toolset to study the brain's innate immune cells. Glia 2020; 68:721-739. [PMID: 31926038 DOI: 10.1002/glia.23781] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 12/21/2019] [Accepted: 12/30/2019] [Indexed: 12/11/2022]
Abstract
Recent advances in the generation of microglia from human induced pluripotent stem cells (iPSCs) have provided exciting new approaches to examine and decipher the biology of microglia. As these techniques continue to evolve to encompass more complex in situ and in vivo paradigms, so too have they begun to yield novel scientific insight into the genetics and function of human microglia. As such, researchers now have access to a toolset comprised of three unique "flavors" of iPSC-derived microglia: in vitro microglia (iMGs), organoid microglia (oMGs), and xenotransplanted microglia (xMGs). The goal of this review is to discuss the variety of research applications that each of these techniques enables and to highlight recent discoveries that these methods have begun to uncover. By presenting the research paradigms in which each model has been successful, as well as the key benefits and limitations of each approach, it is our hope that this review will help interested researchers to incorporate these techniques into their studies, collectively advancing our understanding of human microglia biology.
Collapse
Affiliation(s)
- Jonathan Hasselmann
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, California.,Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, California.,Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, California
| | - Mathew Blurton-Jones
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, California.,Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, California.,Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, California
| |
Collapse
|
10
|
Putz EJ, Eder JM, Reinhardt TA, Sacco RE, Casas E, Lippolis JD. Differential phenotype of immune cells in blood and milk following pegylated granulocyte colony-stimulating factor therapy during a chronic Staphylococcus aureus infection in lactating Holsteins. J Dairy Sci 2019; 102:9268-9284. [PMID: 31400902 DOI: 10.3168/jds.2019-16448] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 06/10/2019] [Indexed: 01/23/2023]
Abstract
Neutrophils are principal host innate immune cell responders to mastitis infections. Thus, therapies have been developed that target neutrophil expansion. This includes the neutrophil-stimulating cytokine granulocyte colony-stimulating factor (gCSF). Pegylated gCSF (PEG-gCSF; Imrestor, Elanco Animal Health, Greenfield, IN) has been shown to reduce the natural incidence of mastitis in periparturient cows in commercial settings and reduce severity of disease against experimental mastitis challenge. Pegylated gCSF stimulates neutrophil expansion but also induces changes in monocyte and lymphocyte circulating numbers, surface protein expression changes, or both. We hypothesized that PEG-gCSF modulates surface expression of monocytes and neutrophils and facilitates their migration to the mammary gland. We challenged 8 mid-lactation Holsteins with approximately 150 cfu of Staphylococcus aureus (Newbould 305) in a single quarter via intramammary infusion. All animals developed chronic infections as assessed by bacteria counts and somatic cell counts (SCC). Ten to 16 wk postchallenge, 4 of the animals were treated with 2 subcutaneous injections of PEG-gCSF 7 d apart. Complete blood counts, SCC, bacterial counts, milk yield, feed intake, neutrophils extracellular trap analysis, and flow cytometric analyses of milk and blood samples were performed at indicated time points for 14 d after the first PEG-gCSF injection. The PEG-gCSF-treated cows had significantly increased numbers of blood neutrophils and lymphocytes compared with control cows. Flow cytometric analyses revealed increased surface expression of myeloperoxidase (MPO) on neutrophils and macrophages in milk but not in blood of treated cows. Neutrophils isolated from blood of PEG-gCSF-treated cows had decreased surface expression of CD62L (L-selectin) in blood, consistent with cell activation. Surprisingly, CD62L cell surface expression was increased on neutrophils and macrophages sourced from milk from treated animals compared with cells isolated from controls. The PEG-gCSF-treated cows did not clear the S. aureus infection, nor did they significantly differ in SCC from controls. These findings provide evidence that PEG-gCSF therapy modifies cell surface expression of neutrophils and monocytes. However, although surface MPO+ cells accumulate in the mammary gland, the lack of bacterial control from these milk-derived cells suggests an incomplete role for PEG-gCSF treatment against chronic S. aureus infection and possibly chronic mammary infections in general.
Collapse
Affiliation(s)
- E J Putz
- Ruminant Diseases and Immunology Research Unit, USDA Agricultural Research Service, National Animal Disease Center, Ames, IA 50010; Oak Ridge Institute for Science and Education, Oak Ridge Associated Universities, Oak Ridge, TN 37830
| | - J M Eder
- Ruminant Diseases and Immunology Research Unit, USDA Agricultural Research Service, National Animal Disease Center, Ames, IA 50010; Immunobiology Interdepartmental Graduate Program, Iowa State University, Ames 50011
| | - T A Reinhardt
- Ruminant Diseases and Immunology Research Unit, USDA Agricultural Research Service, National Animal Disease Center, Ames, IA 50010
| | - R E Sacco
- Ruminant Diseases and Immunology Research Unit, USDA Agricultural Research Service, National Animal Disease Center, Ames, IA 50010; Immunobiology Interdepartmental Graduate Program, Iowa State University, Ames 50011
| | - E Casas
- Ruminant Diseases and Immunology Research Unit, USDA Agricultural Research Service, National Animal Disease Center, Ames, IA 50010
| | - J D Lippolis
- Ruminant Diseases and Immunology Research Unit, USDA Agricultural Research Service, National Animal Disease Center, Ames, IA 50010.
| |
Collapse
|
11
|
Hasselmann J, Coburn MA, England W, Figueroa Velez DX, Kiani Shabestari S, Tu CH, McQuade A, Kolahdouzan M, Echeverria K, Claes C, Nakayama T, Azevedo R, Coufal NG, Han CZ, Cummings BJ, Davtyan H, Glass CK, Healy LM, Gandhi SP, Spitale RC, Blurton-Jones M. Development of a Chimeric Model to Study and Manipulate Human Microglia In Vivo. Neuron 2019; 103:1016-1033.e10. [PMID: 31375314 DOI: 10.1016/j.neuron.2019.07.002] [Citation(s) in RCA: 247] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 06/10/2019] [Accepted: 06/28/2019] [Indexed: 12/19/2022]
Abstract
iPSC-derived microglia offer a powerful tool to study microglial homeostasis and disease-associated inflammatory responses. Yet, microglia are highly sensitive to their environment, exhibiting transcriptomic deficiencies when kept in isolation from the brain. Furthermore, species-specific genetic variations demonstrate that rodent microglia fail to fully recapitulate the human condition. To address this, we developed an approach to study human microglia within a surrogate brain environment. Transplantation of iPSC-derived hematopoietic-progenitors into the postnatal brain of humanized, immune-deficient mice results in context-dependent differentiation into microglia and other CNS macrophages, acquisition of an ex vivo human microglial gene signature, and responsiveness to both acute and chronic insults. Most notably, transplanted microglia exhibit robust transcriptional responses to Aβ-plaques that only partially overlap with that of murine microglia, revealing new, human-specific Aβ-responsive genes. We therefore have demonstrated that this chimeric model provides a powerful new system to examine the in vivo function of patient-derived and genetically modified microglia.
Collapse
Affiliation(s)
- Jonathan Hasselmann
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92696, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92696, USA; Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92696, USA
| | - Morgan A Coburn
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92696, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92696, USA; Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92696, USA
| | - Whitney England
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA
| | - Dario X Figueroa Velez
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92696, USA
| | - Sepideh Kiani Shabestari
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92696, USA
| | - Christina H Tu
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92696, USA
| | - Amanda McQuade
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92696, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92696, USA; Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92696, USA
| | - Mahshad Kolahdouzan
- Neuroimmunology Unit, Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada
| | - Karla Echeverria
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92696, USA
| | - Christel Claes
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92696, USA
| | - Taylor Nakayama
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92696, USA
| | - Ricardo Azevedo
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92696, USA
| | - Nicole G Coufal
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
| | - Claudia Z Han
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA 92093, USA
| | - Brian J Cummings
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92696, USA
| | - Hayk Davtyan
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92696, USA; Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92696, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA 92093, USA; Department of Medicine, University of California, San Diego, San Diego, CA 92093-0651, USA
| | - Luke M Healy
- Neuroimmunology Unit, Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A 2B4, Canada
| | - Sunil P Gandhi
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92696, USA; Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92697, USA
| | - Robert C Spitale
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA; Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92697, USA
| | - Mathew Blurton-Jones
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92696, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92696, USA; Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92696, USA; Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
12
|
MISTRG mice support engraftment and assessment of nonhuman primate hematopoietic stem and progenitor cells. Exp Hematol 2018; 70:31-41.e1. [PMID: 30590092 DOI: 10.1016/j.exphem.2018.12.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 12/20/2022]
Abstract
Preclinical feasibility, safety, and efficacy testing of hematopoietic stem cell (HSC)-mediated gene therapy approaches is commonly performed in large-animal models such as nonhuman primates (NHPs). Here, we wished to determine whether mouse models would allow engraftment of NHP HSPCs, which would enable more facile and less costly evaluation of promising strategies. In this study, we comprehensively tested two mouse strains for the engraftment of NHP CD34+ hematopoietic stem and progenitor cells (HSPCs). No engraftment of NHP HSPCs was observed in NSG mice, whereas the gene-humanized MISTRG model did demonstrate dose-dependent multilineage engraftment of NHP cells in the peripheral blood, bone marrow, spleen, and thymus. Most importantly, and closely mimicking the hematopoietic recovery of autologous stem cell transplantations in the NHP, only HSC-enriched CD34+CD90+CD45RA- cell fractions engrafted and reconstituted the bone marrow stem cell niche in MISTRG mice. In summary, we here report the first "monkeynized" mouse xenograft model that closely recapitulates the autologous hematopoietic reconstitution in the NHP stem and progenitor cell transplantation and gene therapy model. The availability of this model has the potential to pre-evaluate novel HSC-mediated gene therapy approaches, inform studies in the NHP, and improve the overall outcome of large-animal experiments.
Collapse
|
13
|
Powell EJ, Reinhardt TA, Casas E, Lippolis JD. The effect of pegylated granulocyte colony-stimulating factor treatment prior to experimental mastitis in lactating Holsteins. J Dairy Sci 2018; 101:8182-8193. [PMID: 29885891 DOI: 10.3168/jds.2018-14550] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/24/2018] [Indexed: 12/27/2022]
Abstract
Neutrophils are the first-acting and most prominent cellular defense against mastitis-causing pathogens. This makes neutrophil activation and expansion obvious candidates for targeted therapeutics. The granulocyte colony-stimulating factor (G-CSF) cytokine stimulates the bone marrow to produce granulocytes and stem cells and release them into the bloodstream, which results in neutrophilia as well as increasing the presence of other progenitor cells in the bloodstream. A pegylated form of G-CSF (PEG-gCSF) has been shown to significantly decrease naturally occurring mastitis rates in cows postpartum. The use of PEG-gCSF had not been evaluated in response to an experimental mastitis challenge. In an effort to examine the effect and mechanism of PEG-gCSF treatment, we challenged 11 mid-lactation Holsteins with ∼400 cfu Escherichia coli P4 by intramammary infusion. Five cows received 2 PEG-gCSF injections, one at 14 d and the other at 7 d before disease challenge, and 6 cows remained untreated. To evaluate the response of cows to the PEG-gCSF treatment, we measured complete blood counts, somatic cell counts, bacterial counts, milk yield, and feed intake data. The PEG-gCSF-treated cows had significantly increased circulating levels of neutrophils and lymphocytes after each PEG-gCSF injection, as well as following mastitis challenge. The PEG-gCSF-treated cows had significantly lower bacterial counts and lower milk BSA levels at the peak of infection. In addition, control cows had significant decreases in milk yield postinfection and significantly reduced feed intake postinfection compared with PEG-gCSF-treated cows. Collectively, PEG-gCSF treatment resulted in reduced disease severity when administered before a bacterial challenge. Mechanistically, we show that G-CSF treatment increases cell surface expression of an E-selectin ligand before infection on neutrophils and monocytes found in the blood. These cells quickly disappear from the blood shortly after infection, suggesting a mechanism for the reduced mastitis severity by priming immune cells for quick targeting to the site of infection.
Collapse
Affiliation(s)
- E J Powell
- Ruminant Diseases and Immunology Research Unit, USDA Agricultural Research Service, National Animal Disease Center, Ames, IA 50010; Oak Ridge Institute for Science and Education, Oak Ridge Associated Universities, Oak Ridge, TN 37830
| | - T A Reinhardt
- Ruminant Diseases and Immunology Research Unit, USDA Agricultural Research Service, National Animal Disease Center, Ames, IA 50010
| | - E Casas
- Ruminant Diseases and Immunology Research Unit, USDA Agricultural Research Service, National Animal Disease Center, Ames, IA 50010
| | - J D Lippolis
- Ruminant Diseases and Immunology Research Unit, USDA Agricultural Research Service, National Animal Disease Center, Ames, IA 50010.
| |
Collapse
|
14
|
Nieto C, Bragado R, Municio C, Sierra-Filardi E, Alonso B, Escribese MM, Domínguez-Andrés J, Ardavín C, Castrillo A, Vega MA, Puig-Kröger A, Corbí AL. The Activin A-Peroxisome Proliferator-Activated Receptor Gamma Axis Contributes to the Transcriptome of GM-CSF-Conditioned Human Macrophages. Front Immunol 2018; 9:31. [PMID: 29434585 PMCID: PMC5796898 DOI: 10.3389/fimmu.2018.00031] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 01/04/2018] [Indexed: 11/30/2022] Open
Abstract
GM-CSF promotes the functional maturation of lung alveolar macrophages (A-MØ), whose differentiation is dependent on the peroxisome proliferator-activated receptor gamma (PPARγ) transcription factor. In fact, blockade of GM-CSF-initiated signaling or deletion of the PPARγ-encoding gene PPARG leads to functionally defective A-MØ and the onset of pulmonary alveolar proteinosis. In vitro, macrophages generated in the presence of GM-CSF display potent proinflammatory, immunogenic and tumor growth-limiting activities. Since GM-CSF upregulates PPARγ expression, we hypothesized that PPARγ might contribute to the gene signature and functional profile of human GM-CSF-conditioned macrophages. To verify this hypothesis, PPARγ expression and activity was assessed in human monocyte-derived macrophages generated in the presence of GM-CSF [proinflammatory GM-CSF-conditioned human monocyte-derived macrophages (GM-MØ)] or M-CSF (anti-inflammatory M-MØ), as well as in ex vivo isolated human A-MØ. GM-MØ showed higher PPARγ expression than M-MØ, and the expression of PPARγ in GM-MØ was found to largely depend on activin A. Ligand-induced activation of PPARγ also resulted in distinct transcriptional and functional outcomes in GM-MØ and M-MØ. Moreover, and in the absence of exogenous activating ligands, PPARγ knockdown significantly altered the GM-MØ transcriptome, causing a global upregulation of proinflammatory genes and significantly modulating the expression of genes involved in cell proliferation and migration. Similar effects were observed in ex vivo isolated human A-MØ, where PPARγ silencing led to enhanced expression of genes coding for growth factors and chemokines and downregulation of cell surface pathogen receptors. Therefore, PPARγ shapes the transcriptome of GM-CSF-dependent human macrophages (in vitro derived GM-MØ and ex vivo isolated A-MØ) in the absence of exogenous activating ligands, and its expression is primarily regulated by activin A. These results suggest that activin A, through enhancement of PPARγ expression, help macrophages to switch from a proinflammatory to an anti-inflammatory polarization state, thus contributing to limit tissue damage and restore homeostasis.
Collapse
Affiliation(s)
- Concha Nieto
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Rafael Bragado
- Instituto de Investigación Sanitaria, Fundación Jiménez Díaz, Madrid, Spain
| | - Cristina Municio
- Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Elena Sierra-Filardi
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Bárbara Alonso
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - María M Escribese
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Jorge Domínguez-Andrés
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Carlos Ardavín
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Antonio Castrillo
- Instituto Investigaciones Biomédicas "Alberto Sols" (IIBM), and Centro Mixto Consejo Superior de Investigaciones Científicas y Universidad Autónoma de Madrid (ICSIC-UAM), Madrid, Spain.,Unidad de Biomedicina (Unidad Asociada al CSIC), IIBM-Universidad Las Palmas de Gran Canaria (ULPGC), and Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain
| | - Miguel A Vega
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Amaya Puig-Kröger
- Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Angel L Corbí
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| |
Collapse
|
15
|
Riccio A, De Caterina M, Natale D, Grimaldi E, Pronesti G, Montagnani S, Postiglione L. Serum Levels of Granulocyte Macrophage Colony Stimulating Factor (GM-CSF) in a Group of Patients with Systemic Sclerosis. Int J Immunopathol Pharmacol 2016. [DOI: 10.1177/039463209600900102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
In this report we investigate the behaviour of the serum levels of Granulocyte Macrophage Colony Stimulating Factor (GM-CSF) in the course of Systemic Sclerosis (SS). This cytokine is produced mainly by T and NK cells, and its possible role in the pathogenesis of SS has not been previously described in the literature. Serum GM-CSF levels were assayed in 10 female patients, ageing from 35 to 70, affected by SS. These patients were not suffering from other disorders and were not being treated with steroids or immunosuppressive drug. A solid phase immunoenzymatic method was used to assess the serum levels of GM-CSF. Reference values were previously determined in a control group of 36 healthy women blood donors (19 premenopausal and 17 postmenopausal) (x̄=20.1 ±12.3 pg/ml). All the patients but one showed significantly increased serum levels of GM-CSF (x̄= 120.9 ±125.5 pg/ml). The highest levels were found in the two oldest patients, who also had the longest clinical history of SS, but a clear correlation with age, disease duration or clinical manifestations was not evident, even if the postmenopausal age group patients showed a higher mean value of GM-CSF (x̄= 148.0±144.1 pg/ml) than that found in the premenopausal age group (x̄= 57.7±1.4 pg/ml) (in contrast with the findings in the control group). The absence of other pathogenic conditions in our patients suggests that the increase in serum levels of GM-CSF might be linked to the fibroblast proliferation which is typical of SS. However, our results do not explain the role played by this factor in the fibroblastic proliferation process and an in vitro study is necessary to clarify this aspect.
Collapse
Affiliation(s)
- A. Riccio
- Istituto di Medicina Interna e Malattie Dismetaboliche, Università “Federico II” - Facoltà di Medicina e Chirurgia - Napoli
| | - M. De Caterina
- Servizio speciale di Ematologia di Laboratorio, Università “Federico II” - Facoltà di Medicina e Chirurgia - Napoli
| | - D. Natale
- Istituto di Medicina Interna e Malattie Dismetaboliche, Università “Federico II” - Facoltà di Medicina e Chirurgia - Napoli
| | - E. Grimaldi
- Servizio speciale di Ematologia di Laboratorio, Università “Federico II” - Facoltà di Medicina e Chirurgia - Napoli
| | - G. Pronesti
- Istituto di Medicina Interna e Malattie Dismetaboliche, Università “Federico II” - Facoltà di Medicina e Chirurgia - Napoli
| | - S. Montagnani
- Istituto di Anatomia Umana Normale, Università “Federico II” - Facoltà di Medicina e Chirurgia - Napoli
| | - L. Postiglione
- Dipartimento di Biologia e Patologia Cellulare e Molecolare, Italy, Università “Federico II” - Facoltà di Medicina e Chirurgia - Napoli
| |
Collapse
|
16
|
Gallicchio VS, Hughes NK. Effect of G-CSF and M-CSF on the in vitro Toxicity Associated with Zidovudine in Normal Human Bone Marrow Haematopoietic Progenitor Stem Cells. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/095632029300400601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Zidovudine, the antiviral drug used in the treatment of acquired immunodeficiency syndrome (AIDS), causes toxicity to the haematopoietic system. Although use of the haematopoietic growth factors, GM-CSF and erythropoietin have been investigated in clinical trials to modulate antiviral toxicity, there is scant data which supports their ability to ameliorate zidovudine induced toxicity on haematopoietic progenitor cells when combined in vitro. We describe here the results of studies designed to evaluate the capacity of additional haematopoietic factors such as granulocyte-colony stimulating factor (G-CSF) and macrophage-colony stimulating factor (M-CSF) to modulate zidovudine-induced toxicity on G-CSF and M-CSF dependent-colony formation in the presence or absence of zidovudine in vitro. These factors were also studied combined with erythropoietin in culture for the early erythroid progenitor BFU-E using adherent, T-cell, depleted normal human bone marrow cells in the presence or absence of zidovudine. In the presence of zidovudine at the concentration producing 50% inhibition of G- and M-CSF dependent colony formation, (5 × 10−5M), dose-escalation of either G-CSF or M-CSF failed to ameliorate zidovudine toxicity. However, in the presence of zidovudine at the concentration that produces 50% inhibition of BFU-E (5 × 10−9M), and optimal erythropoietin (1 unit ml−1), G-CSF ameliorated zidovudine inhibition of BFU-E, which was not observed with M-CSF. In the presence of erythropoietin, G-CSF increased significantly normal BFU-E. These studies indicate that G-CSF may be useful in ameliorating zidovudine-induced anaemia and suggest G-CSF may act as a synergistic factor to enhance erythropoietin to support the growth of erythroid progenitors in conditions where erythropoitin is ineffective.
Collapse
Affiliation(s)
- V. S. Gallicchio
- Hematology/Oncology Division, Departments of Medicine, Microbiology and Immunology, and Clinical Sciences, Lucille P. Markey Cancer Center, University of Kentucky Medical Center, Lexington, KY 40536-0084, USA
| | - N. K. Hughes
- Department of Veterans Affairs, Lexington, KY 40536-0084, USA
| |
Collapse
|
17
|
Gallicchio VS, Hughes NK, Hulette BC, Noblitt L. Effect of IL-1, IL-6, GM-CSF and Erythropoietin on the in vitro Toxicity Associated with AZT on Human Bone Marrow Haematopoietic Progenitor Stem Cells: CFU-GM and BFU-E. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/095632029100200202] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The drug azidothymidine (AZT), a synthetic thymidine analogue, has been used in the treatment of acquired immunodeficiency syndrome (AIDS). Clinical use of AZT has induced haematopoietic toxicity manifested by anaemia, neutropenia, and overall bone marrow suppression. Cytokines/growth factors, such as erythropoietin (EPO), granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-1 (IL-1), interleukin-6 (IL-6), are agents responsible for the growth and regulation of normal haematopoiesis by influencing various classes of haematopoietic progenitors. We report the results of studies designed to investigate the capacity of these factors to influence the toxicity of AZT. Low density, ≤ 1.077 g/cm3, adherent and/or T-cell depleted normal human marrow cells were co-cultured in the presence or absence of AZT and the appropriate growth factor, i.e. EPO for the early erythroid haematopoietic colony-forming progenitor stem cell (BFU-E) and GM-CSF for the granulocyte–macrophage haematopoietic colony-forming progenitor stem cell (CFU-GM), in dose escalation studies. Additional experiments measured the effect of increasing doses of the cytokines IL-1 and IL-6, alone or in combination in the presence of increasing doses of either EPO or GM-CSF. When comparing the rate of AZT-induced inhibition of BFU-E in vitro, EPO alone (from 2 to 10 U/ml) did not reduce the magnitude of AZT toxicity on BFU-E. GM-CSF alone (up to 1000 U/ml) was ineffective in reversing AZT toxicity on CFU-GM; however, in the presence of either IL-1 and IL-6, AZT toxicity was decreased. These results indicate that certain cytokines/growth factors such as IL-1 or IL-6 in combination with EPO or GM-CSF, but not EPO or GM-CSF alone, may be effective in ameliorating AZT bone marrow toxicity; therefore the use of specific cytokines may be warranted as adjuvant therapy in AIDS.
Collapse
Affiliation(s)
- V. S. Gallicchio
- Hematology/Oncology Division, Departments of Medicine, Microbiology & Immunology, and Clinical, Sciences, Lucille P. Markey Cancer Center, University of Kentucky Medical Center, 800 Rose Street and Veterans Administration Medical Center Lexington, KY 40536–0084, USA
| | - N. K. Hughes
- Hematology/Oncology Division, Departments of Medicine, Microbiology & Immunology, and Clinical, Sciences, Lucille P. Markey Cancer Center, University of Kentucky Medical Center, 800 Rose Street and Veterans Administration Medical Center Lexington, KY 40536–0084, USA
| | - B. C. Hulette
- Hematology/Oncology Division, Departments of Medicine, Microbiology & Immunology, and Clinical, Sciences, Lucille P. Markey Cancer Center, University of Kentucky Medical Center, 800 Rose Street and Veterans Administration Medical Center Lexington, KY 40536–0084, USA
| | - L. Noblitt
- Hematology/Oncology Division, Departments of Medicine, Microbiology & Immunology, and Clinical, Sciences, Lucille P. Markey Cancer Center, University of Kentucky Medical Center, 800 Rose Street and Veterans Administration Medical Center Lexington, KY 40536–0084, USA
| |
Collapse
|
18
|
Large-scale hematopoietic differentiation of human induced pluripotent stem cells provides granulocytes or macrophages for cell replacement therapies. Stem Cell Reports 2015; 4:282-96. [PMID: 25680479 PMCID: PMC4325194 DOI: 10.1016/j.stemcr.2015.01.005] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 01/07/2015] [Accepted: 01/08/2015] [Indexed: 12/15/2022] Open
Abstract
Interleukin-3 (IL-3) is capable of supporting the proliferation of a broad range of hematopoietic cell types, whereas granulocyte colony-stimulating factor (G-CSF) and macrophage CSF (M-CSF) represent critical cytokines in myeloid differentiation. When this was investigated in a pluripotent-stem-cell-based hematopoietic differentiation model, IL-3/G-CSF or IL-3/M-CSF exposure resulted in the continuous generation of myeloid cells from an intermediate myeloid-cell-forming complex containing CD34+ clonogenic progenitor cells for more than 2 months. Whereas IL-3/G-CSF directed differentiation toward CD45+CD11b+CD15+CD16+CD66b+ granulocytic cells of various differentiation stages up to a segmented morphology displaying the capacity of cytokine-directed migration, respiratory burst response, and neutrophil-extracellular-trap formation, exposure to IL-3/M-CSF resulted in CD45+CD11b+CD14+CD163+CD68+ monocyte/macrophage-type cells capable of phagocytosis and cytokine secretion. Hence, we show here that myeloid specification of human pluripotent stem cells by IL-3/G-CSF or IL-3/M-CSF allows for prolonged and large-scale production of myeloid cells, and thus is suited for cell-fate and disease-modeling studies as well as gene- and cell-therapy applications. Myeloid specification of human PSCs by IL-3-/M-CSF, G-CSF, or GM-CSF Large-scale and continuous generation of M2-MΦ or granulocytes by M-CSF or G-CSF Functional iPSC-derived macrophages or granulocytes similar to in-vivo-derived cells
Collapse
|
19
|
Xie J, Zhang C. Ex vivo expansion of hematopoietic stem cells. SCIENCE CHINA-LIFE SCIENCES 2015; 58:839-53. [PMID: 26246379 DOI: 10.1007/s11427-015-4895-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 06/03/2015] [Indexed: 02/03/2023]
Abstract
Ex vivo expansion of hematopoietic stem cells (HSCs) would benefit clinical applications in several aspects, to improve patient survival, utilize cord blood stem cells for adult applications, and selectively propagate stem cell populations after genetic manipulation. In this review we summarize and discuss recent advances in the culture systems of mouse and human HSCs, which include stroma/HSC co-culture, continuous perfusion and fed-batch cultures, and those supplemented with extrinsic ligands, membrane transportable transcription factors, complement components, protein modification enzymes, metabolites, or small molecule chemicals. Some of the expansion systems have been tested in clinical trials. The optimal condition for ex vivo expansion of the primitive and functional human HSCs is still under development. An improved understanding of the mechanisms for HSC cell fate determination and the HSC culture characteristics will guide development of new strategies to overcome difficulties. In the future, development of a combination treatment regimen with agents that enhance self-renewal, block differentiation, and improve homing will be critical. Methods to enhance yields and lower cost during collection and processing should be employed. The employment of an efficient system for ex vivo expansion of HSCs will facilitate the further development of novel strategies for cell and gene therapies including genome editing.
Collapse
Affiliation(s)
- JingJing Xie
- Taishan Scholar Immunology Program, Binzhou Medical University, Yantai, 264003, China
- Departments of Physiology and Developmental Biology, University of Texas Southwestern Medical Center, Dallas, 75390, USA
| | - ChengCheng Zhang
- Departments of Physiology and Developmental Biology, University of Texas Southwestern Medical Center, Dallas, 75390, USA.
| |
Collapse
|
20
|
Hashimoto A, Tanaka T, Itoh Y, Yamagata A, Kitamura N, Tazawa R, Nakagaki K, Nakata K. Low concentrations of recombinant granulocyte macrophage-colony stimulating factor derived from Chinese hamster ovary cells augments long-term bioactivity with delayed clearance in vitro. Cytokine 2014; 68:118-26. [PMID: 24813650 DOI: 10.1016/j.cyto.2014.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 02/18/2014] [Accepted: 03/24/2014] [Indexed: 11/27/2022]
Abstract
To date, the biological activity of granulocyte macrophage-colony stimulating factor (GM-CSF) has been investigated by using mostly Escherichia coli- or yeast cell-derived recombinant human GM-CSF (erhGM-CSF and yrhGM-CSF, respectively). However, Chinese hamster ovary cell-derived recombinant human GM-CSF (crhGM-CSF), as well as natural human GM-CSF, is a distinct molecule that includes modifications by complicated oligosaccharide moieties. In the present study, we reevaluated the bioactivity of crhGM-CSF by comparing it with those of erhGM-CSF and yrhGM-CSF. The effect of short-term stimulation (0.5h) on the activation of neutrophils/monocytes or peripheral blood mononuclear cells (PBMCs) by crhGM-CSF was lower than those with erhGM-CSF or yrhGM-CSF at low concentrations (under 60pM). Intermediate-term stimulation (24h) among the different rhGM-CSFs with respect to its effect on the activation of TF-1 cells, a GM-CSF-dependent cell line, or PBMCs was not significantly different. In contrast, the proliferation/survival of TF-1 cells or PBMCs after long-term stimulation (72-168h) was higher at low concentrations of crhGM-CSF (15-30pM) than that of cells treated with other GM-CSFs. The proportion of apoptotic TF-1 cells after incubation with crhGM-CSF for 72h was lower than that of cells incubated with other rhGM-CSFs. These effects were attenuated by desialylation of crhGM-CSF. Clearance of crhGM-CSF but not desialylated-crhGM-CSF by both TF-1 cells and PBMCs was delayed compared with that of erhGM-CSF or yrhGM-CSF. These results suggest that sialylation of oligosaccharide moieties delayed the clearance of GM-CSF, thus eliciting increased long-term bioactivity in vitro.
Collapse
Affiliation(s)
- Atsushi Hashimoto
- Bioscience Medical Research Center, Niigata University Medical and Dental Hospital, 1-754, Asahimachi-dori, Chuo-ku, Niigata 951-8510, Japan.
| | - Takahiro Tanaka
- Bioscience Medical Research Center, Niigata University Medical and Dental Hospital, 1-754, Asahimachi-dori, Chuo-ku, Niigata 951-8510, Japan.
| | - Yuko Itoh
- Bioscience Medical Research Center, Niigata University Medical and Dental Hospital, 1-754, Asahimachi-dori, Chuo-ku, Niigata 951-8510, Japan.
| | - Akira Yamagata
- Towa Environment Science Co., Ltd. Prophoenix Division, 1-24-22 Nanko-kita, Suminoe, Osaka 559-0034, Japan.
| | - Nobutaka Kitamura
- Department of Medical Informatics, Niigata University Medical and Dental Hospital, 1-754, Asahimachi-dori, Chuo-ku, Niigata 951-8510, Japan.
| | - Ryushi Tazawa
- Bioscience Medical Research Center, Niigata University Medical and Dental Hospital, 1-754, Asahimachi-dori, Chuo-ku, Niigata 951-8510, Japan.
| | - Kazuhide Nakagaki
- Laboratory of Infectious Diseases and Immunology, College of Veterinary Medicine, Nippon Veterinary and Life Science University, 1-1-5, Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan.
| | - Koh Nakata
- Bioscience Medical Research Center, Niigata University Medical and Dental Hospital, 1-754, Asahimachi-dori, Chuo-ku, Niigata 951-8510, Japan.
| |
Collapse
|
21
|
Liu M, Tan H, Zhang X, Liu Z, Cheng Y, Wang D, Wang F. Hematopoietic effects and mechanisms of Fufang e׳jiao jiang on radiotherapy and chemotherapy-induced myelosuppressed mice. JOURNAL OF ETHNOPHARMACOLOGY 2014; 152:575-584. [PMID: 24534527 DOI: 10.1016/j.jep.2014.02.012] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 01/11/2014] [Accepted: 02/08/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fufang e׳jiao jiang (FEJ), which has been widely used in clinic to replenish qi (vital energy) and nourish blood, is a famous traditional Chinese medicine formula made up of Colla corii asini (donkey-hide gelatin prepared by stewing and concentrating from the hide of Equus asinus Linnaeus.), Radix codonopsis pilosulae (the root of Codonopsis pilosula (Franch.) Nannf.), Radix ginseng rubra (the steamed and dried root of Panax ginseng C.A. Mey.), Fructus crataegi (the fruit of Crataegus pinnatifida Bunge) and Radix rehmanniae preparata (the steamed and sun dried tuber of Rehmannia glutinosa (Gaertn.) Libosch. ex Fisch. & C.A. Mey.). The present study aimed to investigate the hematopoietic effects of FEJ on myelosuppressed mice induced by radiotherapy and chemotherapy systematically and to explore the underlying hematopoietic regulation mechanisms. METHODS The myelosuppressed mouse model was induced by (60)Co radiation, cyclophosphamide and chloramphenicol. FEJ was then administered by i.g. at the dosages of 5, 10, or 20 mL/kg·d for 10d. The numbers of blood cells from peripheral blood and bone marrow nucleated cells (BMNC) were counted. Body weight and the thymus and spleen indices were also measured. The numbers of hemopoietic progenitor cells and colony-forming unit-fibroblast (CFU-F) were measured in vitro. The ratio of hematopoietic stem cells (HSC) in BMNC, cell cycle and apoptosis of BMNC were determined by flow cytometry. The histology of femoral bone was examined by H&E staining. The levels of transforming growth factor-β (TGF-β), tumor necrosis factor-α (TNF-α), erythropoietin (EPO), granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-3 (IL-3) and interleukin-6 (IL-6) in serum were measured by ELISA. IL-1β, IL-3, IL-6 mRNA levels in spleen were detected by real-time quantitative PCR (RT-qPCR). In addition, bone marrow stromal cells (BMSC) were cultured in vitro followed by treatment with different doses of FEJ (2.5, 5, 10 μL/mL) for 48 h. Then the levels of cytokines (IL-6, SCF, GM-CSF) in the conditioned media and their mRNA levels in BMSC were determined by ELISA and RT-qPCR, respectively. RESULTS FEJ could significantly increase the numbers of peripheral blood cells and BMNC, and reverse the loss of body weight and the atrophy of thymus and spleen in a dose-dependent manner. The quantities of hemopoietic progenitor cells and CFU-F in bone marrow were also significantly increased in a dose-dependent manner after FEJ administration. A high-dose FEJ of 20 mL/kg·d could significantly increase the ratio of HSC in BMNC, promote bone marrow cells entering the proliferative cycle phase (S+G2/M) and prevent cells from proceeding to the apoptotic phase. FEJ could also improve the femoral bone marrow morphology. Furthermore, FEJ could increase the levels of GM-CSF and IL-3 and reduce the level of TGF-β in serum, and enhance the expressions of IL-1β and IL-3 mRNA in spleen. Lastly, the levels of cytokines (IL-6, SCF, GM-CSF) in the conditioned media and their mRNA levels in BMSC were elevated after treatment with FEJ. CONCLUSIONS FEJ was clearly confirmed to promote the recovery of bone marrow hemopoietic function in a myelosuppressed mouse model, which may be attributed to (i) improving bone marrow hematopoietic microenvironment; (ii) facilitating the cell proliferation and preventing BMNC from apoptosis; (iii) stimulating the expressions of IL-1β, IL-3, IL-6, SCF and GM-CSF and inhibiting the expression of TGF-β.
Collapse
Affiliation(s)
- Maoxuan Liu
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China; National Glycoengineering Research Center, Shandong University, Jinan 250012, China
| | - Haining Tan
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China; National Glycoengineering Research Center, Shandong University, Jinan 250012, China
| | - Xinke Zhang
- Institute of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Zhang Liu
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China; National Glycoengineering Research Center, Shandong University, Jinan 250012, China
| | - Yanna Cheng
- Institute of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Dongliang Wang
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China; Shandong Dong-E-E-Jiao Co. Ltd., Dong׳e 252201, China
| | - Fengshan Wang
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China; National Glycoengineering Research Center, Shandong University, Jinan 250012, China.
| |
Collapse
|
22
|
Wu MF, Chen ST, Hsieh SL. Distinct regulation of dengue virus-induced inflammasome activation in human macrophage subsets. J Biomed Sci 2013; 20:36. [PMID: 23742038 PMCID: PMC3686598 DOI: 10.1186/1423-0127-20-36] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 05/27/2013] [Indexed: 12/27/2022] Open
Abstract
Macrophages (Mϕ) are the major source of inflammatory cytokines and are target cells for dengue virus (DV) replication. However, Mϕ are heterogeneous and their phenotypic and functional diversities are influenced by cytokines that regulate their differentiation, tissue distribution, and defense against invading pathogens. In vitro, human primary macrophages are derived from peripheral blood CD14+ monocytes in the presence of macrophage colony-stimulating factor (M-CSF) or granulocyte macrophage colony-stimulating factor (GM-CSF). These are essential for developing tissue/resting macrophages (M-Mϕ) and inflammatory macrophages (GM-Mϕ), respectively. While IFN production is similar between M-Mϕ and GM-Mϕ, M-Mϕ cannot produce IL-1β after DV infection. In contrast, GM-Mϕ is more susceptible to DV infection and DV triggers CLEC5A in GM-Mϕ to activate NLRP3 inflammasomes, which in turn release IL-18 and IL-1β that are critical for Th17 activation and contribute to disease severity. Thus, GM-Mϕ is more representative than M-Mϕ for investigating inflammasome activation in dengue infection, and is invaluable for revealing the molecular mechanism of pathogen-induced inflammatory reaction. Distinct phenotypes of macrophage subsets under the influence of M-CSF and GM-CSF raise the question of optimal conditions for culturing primary macrophages to study host-pathogen interaction.
Collapse
Affiliation(s)
- Ming-Fang Wu
- Institute of Clinical Medicine & Infection and Immunity Center, National Yang-Ming University, Taipei, 112, Taiwan
| | | | | |
Collapse
|
23
|
Abstract
Humanized mouse models are useful tools to understand pathophysiology and to develop therapies for human diseases. While significant progress has been made in generating immunocompromised mice with a human hematopoietic system, there are still several shortcomings, one of which is poor human myelopoiesis. Here, we report that human CSF-1 knockin mice show augmented frequencies and functions of human myeloid cells. Insertion of human CSF1 into the corresponding mouse locus of Balb/c Rag2(-/-) γc(-/-) mice through VELOCIGENE technology resulted in faithful expression of human CSF-1 in these mice both qualitatively and quantitatively. Intra-hepatic transfer of human fetal liver derived hematopoietic stem and progenitor cells (CD34(+)) in humanized CSF-1 (CSF1(h/h)) newborn mice resulted in more efficient differentiation and enhanced frequencies of human monocytes/macrophages in the bone marrow, spleens, peripheral blood, lungs, liver and peritoneal cavity. Human monocytes/macrophages obtained from the humanized CSF-1 mice show augmented functional properties including migration, phagocytosis, activation and responses to LPS. Thus, humanized mice engineered to express human cytokines will significantly help to overcome the current technical challenges in the field. In addition, humanized CSF-1 mice will be a valuable experimental model to study human myeloid cell biology.
Collapse
|
24
|
|
25
|
Aglietta M, de Felice L, Stacchini A, Sanavio F, Severino A, Simone F, Piacibello W, Mandelli F. Effect of Hemopoietic Growth Factors on the Proliferation of Acute Myeloid and Lymphoid Leukemias. Leuk Lymphoma 2009; 2:207-14. [DOI: 10.3109/10428199009053525] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
26
|
Nagler A, Greenberg PL. Granulocyte-monocyte and Granulocyte-Colony Stimulating Factors in Myelodysplastic Syndromes. Leuk Lymphoma 2009; 5:219-30. [DOI: 10.3109/10428199109068130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
27
|
Franz SE. Erythropoiesis-stimulating agents: development, detection and dangers. Drug Test Anal 2009; 1:245-9. [DOI: 10.1002/dta.51] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
28
|
Harmenberg J, Höglund M, Hellström-Lindberg E. G- and GM-CSF in oncology and oncological haematology. Eur J Haematol Suppl 2009; 55:1-28. [PMID: 7518779 DOI: 10.1111/j.1600-0609.1994.tb01617.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Administration of G- and GM-CSF increases the neutrophil counts in a number of clinical situations. GM-CSF shows the additional effect of increasing the number of monocytes and eosinophil granulocytes. Both G- and GM-CSF affect of neutrophil functions, in the case of GM-CSF there are some potentially negative effects on neutrophil migration and adhesiveness. The clinical relevance of the various effects on mature haematopoietic cells is not fully understood. Clinical data with G-CSF treatment indicate that increased levels of neutrophil granulocytes following cytotoxic chemotherapy may translate into clinical benefit such as a decreased rate of neutropenic infection and an increased cytotoxic chemotherapy dose even though the data are conflicting and the risk of "laboratory cosmetics" is apparent. Regarding treatment with GM-CSF following chemotherapy, the clinical benefit is unclear. The clinical benefit of GM-CSF-induced monocytes and eosinophils is unknown. G- and GM-CSF accelerates neutrophil recovery following autologous or allogeneic BMT. The influence on neutropenic infections is, however, less impressive. Pretreatment with G- or GM-CSF increases the yield of peripheral stem cell harvest, thereby reducing the number of leukaphereses needed. Transplantation of G- and GM-CSF primed autologous peripheral stem cells tends to reduce the period of post-transplant cytopenia, particularly thrombocytopenia, in comparison with traditional ABMT. In patients with MDS, G- and GM-CSF appear to increase the number of neutrophil granulocytes and there is some evidence that patients with severe infectious problems will benefit from this treatment. However, little influence was seen on the main clinical problems with these patients, which are anaemia and thrombocytopenia. In conclusion, G- and GM-CSF are two different proteins with different properties in vivo and in vitro. GM-CSF has, compared with G-CSF, more complex pharmacological effects and a more trouble-some side-effect profile. Early clinical development indicates that both compounds have a substantial influence on the levels of certain blood cells. Whether the increases in different blood cells translate into long-term clinical benefit for greater patient groups is the focus of ongoing research. The effects of G- and GM-CSF may be potentiated by other cytokines, an area which is presently being explored.
Collapse
Affiliation(s)
- J Harmenberg
- Department of Virology, Swedish Institute for Infectious Disease Control, Stockholm
| | | | | |
Collapse
|
29
|
Todo K, Kitagawa K, Sasaki T, Omura-Matsuoka E, Terasaki Y, Oyama N, Yagita Y, Hori M. Granulocyte-macrophage colony-stimulating factor enhances leptomeningeal collateral growth induced by common carotid artery occlusion. Stroke 2008; 39:1875-82. [PMID: 18388343 DOI: 10.1161/strokeaha.107.503433] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Granulocyte-macrophage colony-stimulating factor (GM-CSF) has been reported to accelerate collateral growth (arteriogenesis) at the circle of Willis in rat brain. However, the effect of GM-CSF on leptomeningeal collateral growth has not been established. We examined the effect of unilateral common carotid artery (CCA) occlusion and GM-CSF treatment on leptomeningeal collateral growth in mice. METHODS Adult mice were subjected to unilateral CCA occlusion or sham surgery followed by an alternate-day regimen of GM-CSF (20 microg/kg) or saline injection. On day 7, latex perfusion was performed in 1 set of mice to visualize the leptomeningeal vessels, and the number of Mac-2(+) monocytes/macrophages on the dorsal surface of the brain was counted. In another set of mice, on day 7, permanent ipsilateral middle cerebral artery (MCA) occlusion was performed, and infarct volume was measured. RESULTS Leptomeningeal collateral growth was observed after CCA occlusion, and that was enhanced by GM-CSF treatment. An increase in the number of Mac-2(+) cells on the surface of the brain occurred after CCA occlusion and was enhanced by GM-CSF treatment. Seven days after CCA occlusion, GM-CSF treatment decreased the infarct size attributable to subsequent MCA occlusion. CONCLUSIONS After CCA occlusion, GM-CSF treatment enhanced leptomeningeal collateral growth and decreased the infarct size after MCA occlusion in mice.
Collapse
Affiliation(s)
- Kenichi Todo
- Division of Stroke, Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Vuckovic S, Khalil D, Angel N, Jahnsen F, Hamilton I, Boyce A, Hock B, Hart DNJ. The CMRF58 antibody recognizes a subset of CD123hi dendritic cells in allergen-challenged mucosa. J Leukoc Biol 2004; 77:344-51. [PMID: 15569693 DOI: 10.1189/jlb.1004559] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
CD123(hi) CD11c(-) dendritic cells (CD123(hi) DC) are a distinct subset of human DC present in bone marrow, blood, lymphoid organs, and peripheral tissues. Pathogen stimulation, cytokine, or CD40 ligation induces CD123(hi) DC maturation, involving a shift from their innate immune to cognate antigen-presenting functions. In this study, we revealed that blood CD123(hi) DC in the presence of cytokine (granulocyte macrophage-colony stimulating factor and interleukin-3) undergo progressive, step-wise maturation through an "early" stage, delineated by expression of the antigen detected by the new monoclonal antibody CMRF58 (CD123(hi)CMRF58(+)CD40(-)CD86(-)CD83(-)) to the "late" stage with costimulatory antigen expression (CD123(hi)CMRF58(+)CD40(+)CD86(+)CD83(+/-)). In this early stage, cytokine-maintained CD123(hi) DC do not display changes in their morphology, no longer produce interferon-alpha (IFN-alpha) in response to bacteria, and develop the capacity to induce proliferation and polarization of allogeneic T cells. CD123(hi)CMRF58(+) DC, phenotypically similar to in vitro cytokine-maintained CD123(hi) DC, were not detected in tonsil but are present in allergen-challenged nasal mucosa of allergic individuals. Thus, CD123(hi) DC in certain tissue environments such as allergen-challenged nasal mucosa share a common CD123(hi)CMRF58(+) phenotype with in vitro cytokine-maintained blood CD123(hi) DC characterized by lack of IFN-alpha production.
Collapse
Affiliation(s)
- Slavica Vuckovic
- Mater Medical Research Institute, Aubigny Place, Raymond Terrace, South Brisbane, Queensland 4101, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Rouleau C, Cui K, Feldman L. A functional erythropoietin receptor is necessary for the action of thrombopoietin on erythroid cells lacking c-mpl. Exp Hematol 2004; 32:140-8. [PMID: 15102474 DOI: 10.1016/j.exphem.2003.10.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2003] [Revised: 10/03/2003] [Accepted: 10/13/2003] [Indexed: 10/26/2022]
Abstract
OBJECTIVE We hypothesized that thrombopoietin (TPO) exerts its mitogenic effects on erythroid cells, at least in part, via an interaction of TPO with the cells' erythropoietin receptor (EPO-R). METHODS We used BaF3 cells stably transfected with EPO-R to demonstrate that TPO alone is sufficient to support the long-term growth and proliferation of BaF3/EPO-R cells and to develop a TPO-dependent variant, BaF3/EPO-R(T), which is highly sensitive to and dependent on TPO for its proliferation. Northern analysis and RT-PCR were used to verify that both BaF3/EPO-R and BaF3/EPO-R(T) cells express EPO-R but lack c-mpl, the TPO receptor. To confirm that TPO responsiveness of BaF3/EPO-R(T) is due to TPO's interaction with EPO-R, EPO-R was downregulated by antisense mRNA. RESULTS Downregulation of EPO-R in BaF3/EPO-R(T) cells abolishes responsiveness to both EPO and TPO. Viability of EPO-treated transfectants decreased from 95% to 36%, while that of TPO-treated transfectants decreased from 95% to 9% by 48 hours. Nontransfected BaF3/EPO-R(T), and BaF3/EPO-R(T) transfected with vector alone, remained viable and grew in either EPO or TPO. CONCLUSION Our results suggest a functional EPO-R may be necessary and sufficient for TPO to exert its mitogenic effects on erythroid cells.
Collapse
Affiliation(s)
- Cecile Rouleau
- Laboratory for Cell and Molecular Biology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass 02215, USA
| | | | | |
Collapse
|
32
|
Suzuki H, Katayama N, Ikuta Y, Mukai K, Fujieda A, Mitani H, Araki H, Miyashita H, Hoshino N, Nishikawa H, Nishii K, Minami N, Shiku H. Activities of granulocyte-macrophage colony-stimulating factor and interleukin-3 on monocytes. Am J Hematol 2004; 75:179-89. [PMID: 15054806 DOI: 10.1002/ajh.20010] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We examined the actions of granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-3 (IL-3) on human monocytes, using a serum-free culture system. GM-CSF and IL-3 did not promote the differentiation of monocytes into macrophages but rather into cells with a phenotype compatible with that of immature dendritic cells (DCs). The addition of fetal bovine serum to serum-free cultures with GM-CSF or IL-3 restored the differentiation of monocytes into macrophages. Cells generated with GM-CSF or IL-3 elicited phagocytic activity. Cells generated in the presence of GM-CSF or IL-3, followed by the addition of tumor necrosis factor-alpha, displayed a phenotype of mature DCs, and primed and stimulated immunogenic peptide-specific T lymphocytes. Surprisingly, GM-CSF and IL-3 inhibited macrophage colony-stimulating factor (M-CSF)-dependent differentiation of monocytes into macrophages and induced differentiation into immature DCs. We asked if the inhibition of M-CSF-dependent differentiation into macrophages by GM-CSF or IL-3 was associated with the expression of M-CSF receptors (M-CSFR). GM-CSF or IL-3 down-regulated the expression of M-CSFR. These data demonstrate that GM-CSF and IL-3 primarily support the differentiation of monocytes into DCs and inhibit M-CSF-dependent differentiation into macrophages by suppressing the expression of M-CSFR, thereby promoting differentiation into DCs.
Collapse
Affiliation(s)
- Hirohito Suzuki
- The Second Department of Internal Medicine, Mie University School of Medicine, Mie, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Vargel I, Erdem A, Ertoy D, Pinar A, Erk Y, Altundag MK, Gullu I. Effects of growth factors on doxorubicin-induced skin necrosis: documentation of histomorphological alterations and early treatment by GM-CSF and G-CSF. Ann Plast Surg 2002; 49:646-53. [PMID: 12461449 DOI: 10.1097/00000637-200212000-00015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Extravasation of vesicant antineoplastic agents such as doxorubicin into the skin or subcutaneous tissues may result in loss of the full thickness of the skin or underlying structures. Several treatment methods have been advocated but none has demonstrated any superiority to the others. The authors designed a controlled animal study in 88 rats to test three methods of early treatment of extravasation of the vesicant antineoplastic agent doxorubicin. The first step of the study included 48 Sprague-Dawley rats. All animals received intradermal injections of 1 mg doxorubicin superficially to the panniculus carnosus in the dorsum. The rats were then divided into four groups of 12 rats each, as follows: group 1, no treatment; group 2, immediate intradermal injection of 0.1 ml saline to the same site; group 3, immediate intradermal injection of 10 microg granulocyte macrophage-colony stimulating factor (GM-CSF) in 0.1 ml saline to the same site; group 4, immediate intradermal injection of 10 microg granulocyte-colony stimulating factor (G-CSF) in 0.1 ml saline to the same site. During the next 6 weeks the rats were observed for the development of necrosis. Ulcers developed and reached maximum size two weeks after the injections. The largest ulcers according to area were observed in group 1 and the mean value was 21.25 mm (p < 0.05). Although wound areas were significantly smaller in the saline group than in the control group and the mean value was 7.58 mm (p < 0.05), the smallest lesions were observed in groups 3 and 4, and the mean values were 1.08 mm and 0.83 mm respectively (p < 0.05). There was statistically no difference with regard to mean ulcer area between groups 3 and 4. During the second step of the experiment, the remaining 40 Sprague-Dawley rats were used. Groups containing 10 rats each were designed similarly after all animals received intradermal injections of 1 mg doxorubicin into the back. On the 10th day after the injection, the entire area of the ulcer together with the underlying panniculus carnosus was excised for pathological examination and for determination of glucose 6-phosphate dehydrogenase (G6PD) activity. On microscopic examination, the extravasated ulcer consisted of a large area of ischemic necrosis. There was marked damage to small blood vessels in the form of fibrinoid necrosis and vasculitis. Injured vessel counts were higher in the control group (group 1; p < 0.05). No difference was observed in G6PD activity between the groups. The authors conclude that both saline and tissue growth factors (GM-CSF and G-CSF) are useful for the early treatment of doxorubicin extravasation; however, GM-CSF and G-CSF are more beneficial.
Collapse
Affiliation(s)
- Ibrahim Vargel
- Department of Plastic and Reconstructive Surgery, Hacettepe University School of Medicine, Samanpazari 06100 Ankara, Turkey
| | | | | | | | | | | | | |
Collapse
|
34
|
Okuma E, Inazawa Y, Saeki K, Yuo A. Potential roles of extracellular signal-regulated kinase but not p38 during myeloid differentiation of U937 cells stimulated by cytokines: augmentation of differentiation via prolonged activation of extracellular signal-regulated kinase. Exp Hematol 2002; 30:571-81. [PMID: 12063024 DOI: 10.1016/s0301-472x(02)00801-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To clarify the signaling mechanism of human myeloid differentiation by hematopoietic growth factors and cytokines, we investigated the role of extracellular signal-regulated kinase (ERK) during the differentiation of human monoblastic U937 cells stimulated by granulocyte-macrophage colony-stimulating factor (GM-CSF) and tumor necrosis factor (TNF). MATERIALS AND METHODS Myeloid differentiation was evaluated by morphology, function (respiratory burst activity), and cell surface expression of adhesion molecule (CD11b), and activation of ERK and/or p38 was determined by Western blotting and/or in vitro kinase assay. Inhibition of the ERK pathway was performed using PD98059, a specific inhibitor of this pathway. RESULTS U937 cells were induced to be differentiated by the combination of GM-CSF and TNF, but only minimally by either cytokine alone. Transient phosphorylation and activation of ERK was induced by both GM-CSF alone and combination of the two cytokines, whereas sustained phosphorylation and activation was induced only by the combination. In addition, PD98059, a specific inhibitor of ERK pathway, almost completely abolished this prolonged phosphorylation of ERK and completely blocked differentiation. In contrast, both TNF alone and cytokine combination equivalently phosphorylated p38 in U937 cells, which was dissociated from differentiation, and a specific inhibitor of p38 (SB203580) did not inhibit differentiation. CONCLUSIONS The results indicate potential roles of sustained activation of ERK but not of p38 in the signaling pathways for human myeloid differentiation in U937 cells synergistically stimulated by the two physiologic cytokines GM-CSF and TNF.
Collapse
Affiliation(s)
- Emiko Okuma
- Department of Hematology, Research Institute, International Medical Center of Japan, Tokyo, Japan
| | | | | | | |
Collapse
|
35
|
Ravaud A, Delaunay M, Chevreau C, Coulon V, Debled M, Bret-Dibat C, Courbon F, Gualde N, Nguyen Bui B. Granulocyte-macrophage colony-stimulating factor alone or with dacarbazine in metastatic melanoma: a randomized phase II trial. Br J Cancer 2001; 85:1467-71. [PMID: 11720430 PMCID: PMC2363949 DOI: 10.1054/bjoc.2001.2120] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The potential antitumoral effect of granulocyte-macrophage colony-stimulating factor (GM-CSF) led us to evaluate GM-CSF alone or with dacarbazine (DTIC) in metastatic melanoma in first line randomized phase II. Treatment was arm A: GM-CSF: 5 microg kg(-1), bid, 14 consecutive days every 21 days and arm B: GM-CSF: 5 microg kg(-1), bid, day 2 to day 19 every 21 days and DTIC: 800 mg m(-2), day 1 of each cycle. 32 patients (pts) were included, 15 pts in arm A and 17 in arm B. All pts had visceral metastatic sites. 9 had only one metastatic site. The median number of cycles given was 2 in arm A and 3 in arm B. 100% and 89.4% of the planned dose of GM-CSF was given in arm A and arm B respectively. No objective response was obtained. 19 pts experienced at least WHO grade 3 toxicity. All pts had fever, 29 had a decrease in performance status and 23 had pain. Grade 3 toxicity were fever (38.7%), decrease in performance status (32.3%), pain (19.4%) and dyspnoea (12.5%). In this study, GM-CSF alone or in association with DTIC did not induce any antitumoral activity with subsequent toxicity.
Collapse
Affiliation(s)
- A Ravaud
- Department of Medicine, Institut Bergonié, Regional Cancer Center, Bordeaux, France
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Oren H, Duman N, Abacioglu H, Ozkan H, Irken G. Association between serum macrophage colony-stimulating factor levels and monocyte and thrombocyte counts in healthy, hypoxic, and septic term neonates. Pediatrics 2001; 108:329-32. [PMID: 11483796 DOI: 10.1542/peds.108.2.329] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Macrophage colony-stimulating factor (M-CSF) is a hematopoietic growth factor that mainly stimulates the growth, differentiation, and proliferation of cells of the monocyte-macrophage lineage. There are only limited numbers of studies about M-CSF levels in neonates, but high levels of serum M-CSF have been reported in septic and some thrombocytopenic adult patients. In this study, we investigated the serum M-CSF levels in healthy, septic, and hypoxic term neonates on the first day of life and examined the relationship of serum M-CSF levels and circulating monocyte and thrombocyte counts in these newborn infants. STUDY DESIGN Three groups were defined in this prospective study: group 1, healthy neonates with no risk factors (n = 40); group 2, neonates who had severe hypoxia (n = 20); and group 3, neonates who fulfilled the criteria for early-onset sepsis (n = 18). Blood samples were collected for complete blood cell count and serum M-CSF levels by peripheral venipuncture from each infant in the first 24 hours after birth before any medical therapy. RESULTS The gestational ages and birth weights did not differ significantly between the groups. Serum M-CSF levels of the septic neonates were significantly higher than of both healthy and hypoxic neonates, but did not differ significantly between the healthy and hypoxic neonates. There was no significant correlation between serum M-CSF levels and circulating monocyte counts, but there was a significant inverse correlation between serum M-CSF levels and thrombocyte counts. When this correlation was analyzed according to groups, we determined that this inverse correlation between M-CSF levels and thrombocyte counts was especially significant in the septic neonate group, but not significant in the healthy and hypoxic neonate groups. CONCLUSIONS Serum M-CSF levels are significantly higher in neonates with sepsis. High serum M-CSF levels may have a possible role in the pathogenesis of thrombocytopenia in neonates with sepsis.
Collapse
Affiliation(s)
- H Oren
- Departments of Pediatrics and Microbiology, Dokuz Eylül University Faculty of Medicine, Izmir, Turkey.
| | | | | | | | | |
Collapse
|
37
|
Abstract
Prostate growth and development are primarily under the control of androgens; however, other factors can also influence prostatic growth through alternative pathways. This article discusses some of the major nonandrogenic mediators of prostate growth. Information on the pathways by which these factors exert their effects is also reviewed.
Collapse
Affiliation(s)
- B R Konety
- Department of Urology, University of Pittsburgh, Pennsylvania 15213, USA.
| | | |
Collapse
|
38
|
Correale P, Campoccia G, Tsang KY, Micheli L, Cusi MG, Sabatino M, Bruni G, Sestini S, Petrioli R, Pozzessere D, Marsili S, Fanetti G, Giorgi G, Francini G. Recruitment of dendritic cells and enhanced antigen-specific immune reactivity in cancer patients treated with hr-GM-CSF (Molgramostim) and hr-IL-2. results from a phase Ib clinical trial. Eur J Cancer 2001; 37:892-902. [PMID: 11313178 DOI: 10.1016/s0959-8049(01)00063-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Experimental findings suggest that granulocyte-monocyte-colony stimulating factor (GM-CSF) synergistically interacts with interleukin-2 (IL-2) in generating an efficient antigen-specific immune response. We evaluated the toxicity, antitumour activity and immunobiological effects of human recombinant (hr)-GM-CSF and hr-IL-2 in 25 cancer patients who subcutaneously (s.c.) received hr-GM-CSF 150 microg/day for 5 days, followed by hrIL-2 s.c. for 10 days and 15 days rest. Two of the most common side-effects were bone pain and fever. Of the 24 patients evaluable for response, 3 achieved partial remission, 13 experienced stable disease, and 8 progressed. Cytokine treatment increased the number of monocytes, dendritic cells (DC), and lymphocytes (memory T cells) in the peripheral blood and enhanced the antigen-specific immunoreactivity of these patients. Our results show that the hr-GM-CSF and hr-IL-2 combination is active and well tolerated. Its biological activity may support tumour associated antigen (TAA)-specific anticancer immunotherapy by increasing antigen presenting cell (APC) activity and T cell immune competence in vivo.
Collapse
Affiliation(s)
- P Correale
- Division of Medical Oncology, University of Siena, Viale Bracci 11, 53100, Siena, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Barker CC, Butzner JD, Woodman RC, Parsons HG. Crohn-like enteritis presenting as hypoglycemia in a patient with glycogen storage disease type 1b, treated with granulocyte colony-stimulating factor and splenectomy. J Pediatr Gastroenterol Nutr 2001; 32:197-200. [PMID: 11321392 DOI: 10.1097/00005176-200102000-00019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- C C Barker
- Department of Pediatrics, University of Calgary, Alberta, Canada.
| | | | | | | |
Collapse
|
40
|
Iwasaka H, Kitano T, Miyakawa H, Unoshima M, Shinguu C, Matsumoto S, Noguchi T. Neutrophilia and granulocyte colony-stimulating factor levels after cardiopulmonary bypass. Can J Anaesth 2001; 48:81-4. [PMID: 11212054 DOI: 10.1007/bf03019820] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
PURPOSE The precise mechanism of neutrophilia after cardiac surgery is unknown. Granulocyte colony stimulating factor (G-CSF) can increase the number of leukocytes. The purpose of this study was to evaluate the relationship between serum G-CSF levels and peripheral blood leukocyte counts after cardiac surgery. METHODS We prospectively studied 10 patients undergoing cardiac surgery (coronary artery bypass grafting) using cardiopulmonary bypass (CPB). Plasma G-CSF levels and neutrophil count were measured before induction of anaesthesia, at the end of surgery, and on the first postoperative day. These changes were compared with those in patients undergoing non-cardiac major surgery (control group). RESULTS At the end of surgery, G-CSF levels increased (P < 0.01) in both groups, but were higher in the control than in the cardiac group (3,250 +/- 690 vs 194 +/- 29.5 pg ml(-1), respectively, mean +/- SEM, P < 0.01). On the first postoperative day, G-CSF levels were still high in both groups, and were still higher in the control (710 +/- 179 vs 122 +/- 19.9, respectively, P < 0.01). However, neutrophilia was greater in the cardiac group than in the control. G-CSF response correlated positively with neutrophilia in the control group (r = 0.656, P < 0.05) but not in the cardiac group. CONCLUSIONS Our results indicate that changes in leukocyte count following cardiac surgery are unique to patients undergoing CPB. G-CSF plays an important role as the mediator of neutrophilia after non-cardiac surgery, but not after cardiac surgery with CPB.
Collapse
Affiliation(s)
- H Iwasaka
- Department of Anaesthesiology, Oita Medical University, Japan.
| | | | | | | | | | | | | |
Collapse
|
41
|
Schlemmer R, Spruss T, Bernhardt G, Schönenberger H. Does [meso-1,2-bis(2,6-dichloro-4-hydroxyphenyl)ethylenediamine]- dichloro-platinum(II) act as an immune response modifier? Part III: Progressively growing MXT-M-3,2 breast cancer stimulates the proliferation of phagocytes in B6D2F1 mice. Arch Pharm (Weinheim) 2000; 333:404-14. [PMID: 11199470 DOI: 10.1002/1521-4184(200012)333:12<404::aid-ardp404>3.0.co;2-m] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
MXT-M-3,2 breast cancer implanted into female B6D2F1 mice accelerates the growth of an identical second tumor. This process is accompanied by a significant increase of the granulocyte and monocyte numbers in the blood and of the granulocyte and macrophage numbers in the spleen. A significant positive correlation of strong intensity was found between the tumor weight on the one hand and the number of the granulocytes and macrophages on the other hand. The tumor-dependent promotion of the myelopoiesis is explained with a secretion of hematopoietic growth factors, e.g. of the granulocyte-macrophage-stimulating growth factor (GM-CSF), by the breast cancer cells.
Collapse
Affiliation(s)
- R Schlemmer
- Lehrstuhl für Pharmazeutische Chemie II, Institut für Pharmazie, Universität Regensburg, D-93040 Regensburg, Germany
| | | | | | | |
Collapse
|
42
|
Wolach B, Gavrieli R, Pomeranz A. Effect of granulocyte and granulocyte macrophage colony stimulating factors (G-CSF and GM-CSF) on neonatal neutrophil functions. Pediatr Res 2000; 48:369-73. [PMID: 10960505 DOI: 10.1203/00006450-200009000-00018] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Although there are many studies on the effect of granulocyte and granulocyte-macrophage colony stimulating factors (G/GM-CSF) on adult neutrophil functions, there is little information regarding their influence on neonatal cells. We studied the in vitro effect of G/GM-CSF on neutrophil chemotaxis, polarization, and superoxide anion generation in 47 neonates compared with 35 adults. We found that G-CSF and GM-CSF significantly enhanced the chemotaxis of newborn infants' neutrophils, normalizing their chemotactic defect [from 35 +/- 7 cells/field (mean +/- SE) to 49 +/- 5 cells/field with G-CSF, p < 0.05 and to 55 +/- 4 cells/field with GM-CSF, p < 0.001]. It is notable that the maximal neutrophil response to the cytokines was observed particularly in the newborn infants with severe impairment in their chemotactic activity. Statistical analysis of the data showed a significant inverse correlation, which supported this observation (r = -0.6, p < 0.02 for G-CSF; r = -0.76, p < 0.001 for GM-CSF). The reduced polarization of neonatal compared with adult cells [71 +/- 5% versus 86 +/- 2% (mean +/- SE), p < 0.05], was corrected by CSF-priming (to 87 +/- 4% with G-CSF and to 92 +/- 2% with GM-CSF, p < 0.05). In addition, the neutrophil superoxide generation was significantly improved in both groups following the CSF-priming. GM-CSF and G-CSF gave comparable results in all functions studied except that GM-CSF improved superoxide release to a greater extent. This study shows a significant improvement of the neonatal neutrophil functions following in vitro CSF-priming and contributes to a better understanding of the neonatal neutrophil behavior when treated with G/GM-CSF.
Collapse
Affiliation(s)
- B Wolach
- Department of Pediatrics, the Pediatric Hematology Clinic, Meir General Hospital, Kfar-Sava, Israel
| | | | | |
Collapse
|
43
|
Inukai T, Sugita K, Mitsui K, Iijima K, Goi K, Tezuka T, Kojika S, Kagami K, Mori T, Kinoshita A, Suzuki T, Okazaki-Koyama T, Nakazawa S. Participation of granulocyte colony-stimulating factor in the growth regulation of leukemia cells from Philadelphia chromosome-positive acute leukemia and blast crisis of chronic myeloid leukemia. Leukemia 2000; 14:1386-95. [PMID: 10942233 DOI: 10.1038/sj.leu.2401837] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Granulocyte colony-stimulating factor (G-CSF) has been shown to support the growth of multipotential hematopoietic stem cells in addition to the cells of neutrophilic lineage. Philadelphia chromosome (Ph1)-positive leukemia has its origin in the hematopoietic stem cell. In the present study, we demonstrated that the proliferation of leukemic cells from chronic myeloid leukemia in blast crisis (CML-BC) and Ph1-positive acute lymphoblastic leukemia (ALL) cases is frequently stimulated with G-CSF in vitro. We next studied a total of 12 leukemic cell lines established from CML-BC (n= 6) and Ph1-positive acute leukemia (n= 6): four 'myeloid', five 'biphenotypic', and three 'lymphoid' types. All cell lines expressed G-CSF receptor (G-CSFR) in flow cytometric analysis, but their proliferative response to G-CSF in 3H-thymidine incorporation assay varied. The 'biphenotypic' cell lines expressed G-CSFR at higher levels and showed the most pronounced response to G-CSF. The 'lymphoid' cell lines showed intermediate G-CSFR expression with the modest response to G-CSF. Unexpectedly, 'myeloid' cell lines showed lower G-CSFR expression and lower G-CSF response compared with 'biphenotypic' cell lines. In three of four 'myeloid' cell lines, proliferation was partially inhibited by an addition of anti-G-CSF neutralizing monoclonal antibody into culture medium. Further, the % inhibition of 3H-thymidine uptake of cell lines positively correlated with the amount of their intracellular G-CSF measured by enzyme immunoassay, suggesting an autocrine growth mechanism via the G-CSF/G-CSFR interaction. These results suggest that G-CSF play an important role in the growth regulation of leukemia cells from Ph1-positive acute leukemia and CML-BC.
Collapse
Affiliation(s)
- T Inukai
- Department of Pediatrics, Yamanashi Medical University, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Yao GQ, Sun BH, Insogna KL, Weir EC. Nuclear factor-kappaB p50 is required for tumor necrosis factor-alpha-induced colony-stimulating factor-1 gene expression in osteoblasts. Endocrinology 2000; 141:2914-22. [PMID: 10919279 DOI: 10.1210/endo.141.8.7592] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Colony-stimulating factor (CSF)-1 is a hematopoietic growth factor that is released by osteoblasts and is recognized to play a critical role in bone remodeling in vivo and in vitro. We have reported that osteoblasts express CSF-1 constitutively and that tumor necrosis factor (TNF)-alpha, a potent bone-resorbing agent, increases CSF-1 gene expression by a transcriptional mechanism. In the present study, we report that an NF-kappaB site in the CSF-1 promoter is required for TNF-alpha-induced CSF-1 expression in osteoblasts. As determined by electrophoretic mobility shift assays, antiserum against the NF-kappaB-binding protein, p50, retarded the mobility of the inducible complex, whereas antisera against p52, p65, c-Rel, Rel B, IkappaB alpha, IkappaB gamma, and Bcl-3 had no effect. To further confirm that p50 is necessary for TNF-alpha-induced CSF-1 expression in osteoblasts, CSF-1 messenger RNA expression from untreated and TNF-alpha-treated osteoblasts, prepared from wild-type and p50 knock-out mice, was examined by Northern analysis. CSF-1 messenger RNA was increased by TNF treatment in wild-type mice but not in NF-kappaB p50 knock-out mice. Our findings support the conclusion that the NF-kappaB subunit p50 is critical for TNF-induced CSF-1 expression in osteoblasts.
Collapse
Affiliation(s)
- G Q Yao
- The Section of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut 06520-8016, USA.
| | | | | | | |
Collapse
|
45
|
Abstract
BACKGROUND Vascular endothelium represents a complex network of cells producing a large number of active substrates affecting physiologic, metabolic, and immunologic properties of the whole organism, as well as particular organs or tissues. The potential influence of endothelium-derived paracrine factors on prostate cancer progression has only begun to be examined. METHODS This review summarizes recent literature on endothelium-derived factors, including vasoactive agents, peptide growth factors, cytokines, and colony-stimulating factors, involved in the development and progression of prostate cancer. RESULTS Endothelial cells produce an array of active substrates, many of which have been shown to influence prostate cancer growth. Available data demonstrate the positive impact of such molecules as endothelin-1, basic FGF, TGF-beta, IL-6, and IL-8 on prostate cancer progression. Many other endothelium-derived factors NO, IGF, PDGF, IL-1, G-CSF, and GM-CSF (Nitric Oxide, Insulin-Like Growth Factor, Platelet-Derived Growth Factor, Interleukin-1, Granulocyte Colony Stimulating Factor, and Granulocyte-Macrophage Colony Stimulating Factor) are, at best, implicated in prostate cancer growth, and in most cases support cancer progression. CONCLUSIONS A better understanding of endothelium-derived factors, as paracrine mediators of prostate carcinogenesis and progression, should aid in the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- G Pirtskhalaishvili
- Department of Urology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15213, USA
| | | |
Collapse
|
46
|
Okuma E, Saeki K, Shimura M, Ishizaka Y, Yasugi E, Yuo A. Induction of apoptosis in human hematopoietic U937 cells by granulocyte-macrophage colony-stimulating factor: possible existence of caspase 3-like pathway. Leukemia 2000; 14:612-9. [PMID: 10764146 DOI: 10.1038/sj.leu.2401716] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) induced apoptosis in human hematopoietic U937 cells by itself and in a synergistic manner with tumor necrosis factor (TNF). GM-CSF-induced apoptosis was not inhibited by caspase inhibitors YVAD-CMK, DEVD-CHO and z-VAD-FMK, under the condition that these inhibitors potently suppressed TNF-induced apoptosis. Both GM-CSF and TNF induced caspase 3-like activity in this cell line though the time course was distinct between two cytokines, and combined stimulation of cells with GM-CSF plus TNF induced additive or synergistic activation of caspase 3-like activity. Amount of immunoreactive cleaved forms of caspase 3 recognized by specific antibody was completely dissociated with its enzymatic activity when the cells were stimulated with GM-CSF, but not with TNF. These results indicate that GM-CSF induces apoptosis of U937 cells via unknown pathway, which seems to be mediated by caspase 3-like activity, yet not caspase 3 itself, resistant to the caspase inhibitors, and synergistically interacts with conventional caspase 3 pathway of TNF. Possible involvement of caspases 1 and 8 (-like activity) but not caspase 7 in this pathway was also suggested.
Collapse
Affiliation(s)
- E Okuma
- Department of Hematology, Research Institute, International Medical Center of Japan, Tokyo
| | | | | | | | | | | |
Collapse
|
47
|
Strelkov LA, Mikhailova AA, Fonina LA, Petrov RV. A new endogenous differentiating factor (myelopeptide-4) for myeloid cells. FEBS Lett 2000; 470:281-4. [PMID: 10745082 DOI: 10.1016/s0014-5793(00)01344-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Along with known lymphokines involved in the regulation of hematopoiesis, a new differentiating factor (myelopeptide-4, MP-4) for myeloid cells was found. The peptide (Phe-Arg-Pro-Arg-Ile-Met-Thr-Pro) originally isolated from the culture medium of porcine bone marrow cell culture was examined for its ability to induce differentiation in two human myeloid leukemia cell lines, HL-60 and K-562. Agents with well-known differentiation-inducing activity, such as phorbol myristate acetate, dimethylsulfoxide and the lymphokines were used as a reference. It has been shown that MP-4 significantly influences the integral characteristics of metabolism, expression of surface antigens and morphology of these cells. It decreased the level of chromosomal DNA synthesis and, in parallel, increased the total protein synthesis in both HL-60 and K-562 cells. MP-4 induced the expression of CD14 monocyte-specific surface antigen and the appearance of mature monocytes/macrophages in HL-60 cell cultures. There was a good correlation of cell metabolic/morphological changes and the CD14 marker expression for HL-60 cells. A similar phenomenon was observed in K-562 cells treated with MP-4 when the levels of hemoglobin synthesis were detected in their cytoplasm. Thus, we consider MP-4 as a new endogenous differentiating factor for myeloid cells.
Collapse
Affiliation(s)
- L A Strelkov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 16/10 Miklukho-Maklaya, 117871, Moscow, Russia.
| | | | | | | |
Collapse
|
48
|
Yano S, Hanibuchi M, Nishioka Y, Nokihara H, Nishimura N, Tsuruo T, Sone S. Combined therapy with anti-P-glycoprotein antibody and macrophage colony-stimulating factor gene transduction for multiorgan metastases of multidrug-resistant human small cell lung cancer in NK cell-depleted SCID mice. Int J Cancer 1999; 82:105-11. [PMID: 10360828 DOI: 10.1002/(sici)1097-0215(19990702)82:1<105::aid-ijc18>3.0.co;2-c] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Our aim was to determine the antimetastatic potential of anti-P-glycoprotein (P-gp) antibodies (Abs) against multidrug-resistant (MDR) human small cell lung cancer (SCLC) cells expressing P-gp. Human SCLC cells H69 (P-gp negative) and its etoposide-resistant variant H69/YP (P-gp positive) were used. H69 and H69/VP cells injected i.v. metastasized to the liver, kidneys and systemic lymph nodes of NK cell-depleted severe combined immunodeficient (SCID) mice. H69/VP cells, but not H69 cells, were resistant to treatments with vindesine. Treatment with mouse-human chimeric anti-P-gp Ab (MH162) and its mouse counterpart (MRK-16) reduced metastasis of H69/VP cells in various organs and prolonged the survival of tumor-bearing mice, although they were less effective if injected at late times (after 28 days). Treatment with another mouse anti-Pgp Ab, MRK-17, was effective only against liver metastasis. MH162 and MRK-16 efficiently induced Ab-dependent cellular cytotoxicity (ADCC) by peritoneal macrophages against H69/VP cells in vitro, but MRK-17 was less effective, in accordance with their in vivo antimetastatic potential. Gene transfection of macrophage colony-stimulating factor (M-CSF) into H69/VP cells to augment macrophage-mediated ADCC resulted in inhibition of metastasis to the liver and lymph nodes, but not kidneys. Combined treatment with a low dose of MRK-16 completely cured metastasis of M-CSF transfectant, but not of the mock transfectant. Our findings suggest that while anti-P-gp Abs had antimetastatic potential against SCLC cells expressing P-gp, combined treatment with M-CSF gene transduction to augment the therapeutic efficacy of anti-P-gp Abs may be beneficial for eradicating metastatic MDR SCLC in humans.
Collapse
Affiliation(s)
- S Yano
- Third Department of Internal Medicine, University of Tokushima School of Medicine, Japan
| | | | | | | | | | | | | |
Collapse
|
49
|
|
50
|
Postiglione L, Montagnani S, Riccio A, Ladogana P, Salzano S, Vallefuoco L, Rossi G. Expression of GM-CSF receptor and "in vitro" effects of GM-CSF on human fibroblasts. Life Sci 1998; 63:327-36. [PMID: 9714420 DOI: 10.1016/s0024-3205(98)00281-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In the present study the effects of Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF) on fibroblast growth and activity have been studied. In this regard the AA have evaluated in primary cultures of human gengival normal fibroblasts (PG1 cells): a)-the expression of GM-CSF receptor (GM-CSFR) (alfa unit) on the cell surface; b)-the in vitro effects of different doses of GM-CSF on the GM-CSFR expression and on the proliferation and activity of fibroblasts. PG1 cells have been stimulated in vitro with different concentrations of GM-CSF (10, 50, 80, 100 and 150 ng/ml) using promonocytic cell line U937 as positive control for GM-CSFR expression. GM-CSFR was investigated by flow cytometry, with mouse monoclonal antibody (mAb) against the alfa chain of the human GM-CSFR and fluorescein-conjugated goat antimouse immunoglobulin G (IgG). At high GM-CSF concentration (80 ng/ml) the AA observed: 1)-A marked increase of GM-CSFR expression evaluated as fluorescence intensity (about three fold in respect to the controls); 2)-Maximal increase of PG1 cells proliferation. Moreover immunofluorescence on fibroblasts obtained from culture plates showed increased actin stress fibers and fibronectin production with low stimulation by GM-CSF, while higher concentration of this cytokine determined increased proliferation of cells, but a decreased formation of actine fibers and vinculin plaques. These results demonstrate: 1)-The presence of GM-CSFR on the surface of fibroblasts; 2)-The proliferation and the synthesis activity of these cells (in vitro) are modulated by different concentration of GM-CSF. We hypothesize that GM-CSF until 80 ng/ml can upregulate the expression of the receptor. Therefore, on the basis of previous findings of high serum levels of GM-CSF in course of scleroderma, a disease characterized by fibroblast hyperactivity, a possible role of this cytokine in the pathogenic process of this disease can be hypothesized.
Collapse
Affiliation(s)
- L Postiglione
- Dip. di Biologia e Patologia Molecolare e Cellulare, Università degli Studi Federico II, Naples, Italy
| | | | | | | | | | | | | |
Collapse
|